EP1977245B1 - Method for determining the concentration of virus particles/virus antigens - Google Patents

Method for determining the concentration of virus particles/virus antigens Download PDF

Info

Publication number
EP1977245B1
EP1977245B1 EP07702744.9A EP07702744A EP1977245B1 EP 1977245 B1 EP1977245 B1 EP 1977245B1 EP 07702744 A EP07702744 A EP 07702744A EP 1977245 B1 EP1977245 B1 EP 1977245B1
Authority
EP
European Patent Office
Prior art keywords
virus
antigens
sample
particles
concentration
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP07702744.9A
Other languages
German (de)
French (fr)
Other versions
EP1977245A1 (en
Inventor
Holger Kost
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis Vaccines and Diagnostics Inc
Original Assignee
Novartis Vaccines and Diagnostics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Vaccines and Diagnostics Inc filed Critical Novartis Vaccines and Diagnostics Inc
Priority to EP07702744.9A priority Critical patent/EP1977245B1/en
Publication of EP1977245A1 publication Critical patent/EP1977245A1/en
Application granted granted Critical
Publication of EP1977245B1 publication Critical patent/EP1977245B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q3/00Condition responsive control processes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/04Determining presence or kind of microorganism; Use of selective media for testing antibiotics or bacteriocides; Compositions containing a chemical indicator therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/01DNA viruses
    • G01N2333/065Poxviridae, e.g. avipoxvirus
    • G01N2333/07Vaccinia virus; Variola virus
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/11Orthomyxoviridae, e.g. influenza virus

Definitions

  • the invention provides a method for determining the concentration of virus particles/virus antigens in a sample.
  • the invention relates to determining the concentration of influenza virus particles/influenza virus antigens in a sample.
  • the invention relates to the use of a cation exchange matrix for the determination of the concentration of virus particles/virus antigens in a sample.
  • influenza vaccines can be derived from virus propagation cultures which make use of embryonated chicken eggs. In these cultures, the virus is isolated from the allantoic fluid of infected eggs and the antigens are used as a vaccine either in the form of complete or disintegrated virus particles.
  • viruses such as influenza, rabies, mumps, measles, rubella and tick-borne encephalitis virus may also be replicated by use of specific mammalian cell lines.
  • the host system, culturing conditions and the virus to be propagated must be adapted to one another.
  • the primary virus yield of a virus propagation culture can considerably vary between different batches or production runs.
  • the determination of primary yield of an influenza virus propagation culture is performed, for example, by measuring the haemagglutinin (HA) titer.
  • HA haemagglutinin
  • the aggregation of hen erythrocytes is used to indicate the amount of virus-derived haemagglutinin in the cell-free virus preparations, such as a crude sample aliquot of a given culture.
  • an assessment of the HA amount can be made.
  • the amount of haemagglutinin is indicative for the number of viral particles in the sample.
  • this method is strain-specific and, moreover, only semiquantitative. In fact, only factor 2-steps in sensitivity can be determined.
  • influenza virus yield can be assessed by employing the well-accepted single radial immunodiffusion (also referred to in literature as SRID or SRD test).
  • US2003/0170710 describes a method of purifying and quantifying adenovirus by anion-exchange chromatography.
  • US4,724,210 teaches a method for the purification of influenza viruses.
  • An important drawback of the above methods as presently used in the field resides in the fact that evaluation of the primary virus yield of a given virus propagation culture according to the above methods can only be reliably performed after having separated the virus from the culture medium.
  • purification of virus from large propagation cultures is both cost-intensive and time-consuming. Presently, it is not possible to evaluate the amount of virus in a given virus propagation culture within a period of several hours or less.
  • the determination method should provide a result so rapidly that such result can be used as a basis for deciding whether or not the virus propagation culture is further processed and subjected to elaborate purification methods.
  • the present invention solves this problem and provides additional benefits as well.
  • the present invention provides a method for determining the concentration of viral particles and/or antigens in a virus-containing sample within a short period of time as defined in the claims.
  • the virus-containing sample can be a crude sample of a virus propagation culture, such as a culture of MDCK cells, and the method according to the invention is performed with an aliquot of said culture.
  • examination of the aliquot can be performed without the necessity to subject the complete culture volume to downstream purification.
  • the invention provides a method for determining the concentration of virus particles/virus antigens in a sample comprising the steps of
  • the virus particles and/or virus antigens in the sample are bound to a cation-exchange matrix in a first step.
  • a cation-exchange matrix can readily be purchased from several manufacturers, such as SP SepharoseTM XL from GE Healthcare Life Sciences, Sartobind S from Sartorius, and the like.
  • the column used in the method according to the invention comprises a cellulose matrix.
  • the cellulose matrix has a gel exclusion limit of 2000 to 4000 Dalton, more preferably 3000 Dalton.
  • the cellulose matrix preferably comprises activated groups.
  • Preferred activated groups include, for example, sulfate ester or other weak cation exchange groups, such as Carboxymethyl (CM).
  • CM Carboxymethyl
  • the cellulose matrix comprises sulfate ester as activated groups.
  • the sulfate esters are present on the cellulose matrix in a comparatively low concentration.
  • the matrix can comprise 500-900, preferably 700 ⁇ g sulfur per gram dry matrix material.
  • a cellulose matrix which is particularly suitable for a method according to the invention can be purchased from Millipore GmbH, Eschborn, Germany, under the trademark Matrex ® cellufine TM Sulfate.
  • the sample containing the virus particles and/or virus antigens is contacted with the cation-exchange matrix under low salt conditions that allow binding of said virus particles and/or virus antigens to said cation-exchange matrix.
  • low salt conditions are conditions of low ionic strength, i.e. binding of the virus particles and/or virus antigens in step a) is effected under low salt conditions.
  • contacting and binding is performed in a suitable buffer solution.
  • the choice of the buffer is not critical, so that most buffer solutions commonly used for ion-exchange chromatography may be used.
  • the buffer solution used for contacting the virus particles and/or virus antigens to the matrix can comprise NaCl, Na 2 HPO 4 , NaHPO 4 , Na 3 PO 4 , Na 3 SO 4 , KCl, K 2 HPO 4 , KHPO 4 , K 3 PO 4 , K 2 SO 4 , MgCl 2 , NH 4 Cl, and the like.
  • the total salt concentration of the sample can be up to 0,1 mol/l, for example, 0,01 mol/l, or 0,05 mol/l.
  • the material comprising the virus particles and/or virus antigens for example an aliquot of a virus propagation culture
  • the material comprising the virus particles and/or virus antigens may be desalted before applying the sample to ion-exchange chromatography.
  • contacting of the virus particles and/or virus antigens in step a) is effected in the presence of a buffer solution comprising Na 2 HPO 4 .
  • the concentration of Na 2 HPO 4 in the sample is between 0,01 and 0,1 mol/l. Most preferably, the concentration of Na 2 HPO 4 in the sample is 0,05 mol/l.
  • Contacting the sample with the matrix can be performed in a pH range of about 6,0 to 8,0, preferably 7,0 to 7,8. Most preferably, contacting is performed at a pH of 7,5. To get quantitative results, one has to ensure that the binding capacity of the respective matrix is by far higher than the amount of virus particles and/or virus antigens in the sample to be examined. If the concentration of virus particles and/or virus antigens is too high, dilution should be considered.
  • virus particles and/or virus antigens After the virus particles and/or virus antigens have been bound to the cation-exchange matrix, several washing steps may be performed in order to separate contaminating components of the sample, for example cellular debris originating from host cells used for virus propagation or medium components, from the virus particles and/or virus antigens.
  • Washing can be performed according to common methods used in the field of chromatography using well known buffers of low ionic strength.
  • buffers of low ionic strength For an overview of numerous chromatography methods and techniques, reference is made to " Ion Exchange Chromatography, Principles and Methods", Amersham Pharmacia Edition AA (1998 ).
  • the same buffer which has been used for contacting the virus particles and/or virus antigens to the ion-exchange matrix is used for washing.
  • a sodium phosphate buffer such as Na 2 HPO 4 , having a concentration of 10 to 100 mmol/l for washing.
  • the buffer may have a concentration of 20, 40, 60, 80, or 100 mmol/l.
  • the Na 2 HPO 4 buffer has a concentration of 50 mmol/l.
  • the washing buffer may contain several other components, such as further buffering agents like TRIS (Tris(hydroxymethyl)-aminomethan), MOPS (3-(N-morpholino)-Propansulfonklare) or HEPES (N-(2-Hydroxyethyl)-piperazin-N'-ethansulfonklare) or detergents like Triton-X 100, sodium dodecyl sulfate, polysorbate, polyoxyethylene sorbitol esters such as polysorbate 80 (Tween-80) or polysorbate 20 (Tween-20), polyoxypropylene-polyoxyethylene esters such as poloxamer 188, polyoxyethylene alcohols such as BRIJ 35, and the like.
  • TRIS Tris(hydroxymethyl)-aminomethan
  • MOPS 3-(N-morpholino)-Propansulfonklare
  • HEPES N-(2-Hy
  • virus particles and/or virus antigens are eluted from the column by use of a buffer containing substances which interfere with the interaction of the virus particles and/or virus antigens with the cation-exchange matrix.
  • Such buffers may comprise the same salts as the buffers used for binding the virus particles and/or virus antigens to the column (see above), however in higher concentrations.
  • elution of the virus particles and/or virus antigens in step c) is effected under high salt conditions.
  • the concentration of the buffer used for elution depends on the nature of the buffer.
  • the salt concentration of the elution buffer may range from about 0,8 to 5 mol/l.
  • the salt concentration of the elution buffer may be 1, 1,2, 1,5, 1,8, 2, 2,2, 2,5, 2,8, 3, 3,5 4, or 4,5 mol/l.
  • eluting the virus particles and/or virus antigens in step c) is effected in the presence of a NaCl buffer.
  • the NaCl buffer preferably has a concentration of between 1,0 and 2,0 mol/l, more preferably 1,2 mol/l.
  • an NH 4 SO 4 buffer having a concentration of between 1,0 and 2,0 mol/l can be used for elution. Elution can also be performed in two or more steps using buffers of different concentrations.
  • the elution is performed in two steps, wherein a first elution is performed by use of a sodium chloride buffer of 1,2 mol/l, and subsequently, a second elution is performed by use of a sodium chloride buffer of 3 mol/l. It will be understood by those skilled in the art that elution can also be performed by a linear gradient with increasing salt concentrations.
  • the concentration of both complete virus particles and/or specific parts thereof, such as surface proteins or other protein components can be determined.
  • the method of the invention is particularly suitable to determine the concentration of complete virus particles, such as influenza virus particles, in a sample.
  • the method provides for determining the yield of a virus propagation culture, such as a MDCK cell culture which has been infected with a virus, such as an influenza virus.
  • a virus propagation culture such as a MDCK cell culture which has been infected with a virus, such as an influenza virus.
  • the present method is also useful for determining their concentration.
  • both the concentration of complete virus particles and virus antigens can be determined in parallel, provided that they elute with different retention times.
  • the virus-related substances eluting from the column are detected by measuring the UV absorbance of the eluting substances at a specific wave length.
  • the choice of the wave length depends on the nature of the virus to be detected.
  • virus particles such as influenza virus particles can be detected in a range of 210 to 350 nm, for example 280 nm.
  • the eluting substances are detected by absorbance measurement at a wave length of 280 nm.
  • common UV detection means which are well known in the field of chromatography may be used.
  • the virus particles and/or virus antigens to be determined according to the method of the present invention are HCV, measles virus, HBV, vaccinia virus, lassa virus, HSV-1, HSV-2, yellow fever virus, rubella virus, HIV and influenza virus.
  • the virus particles and/or virus antigens originate from an influenza virus.
  • the influenza virus may be selected, for example, from the strains A/New Caledonia H1N1, B/Jingsu, A/Wyoming H3N2, A7New York H3N2, B/Malaysia and other strains.
  • the sample, which comprises the virus particles and/or virus antigens preferably can be an aliquot of a virus propagation culture.
  • virus propagation culture means any culture which serves the purpose of propagating viruses or virus antigens.
  • the term in particular includes any kind of cell or tissue culture, in which the cells or tissues, for example vertebrate cells, are used as a host for the replication of the virus.
  • the aliquot may be taken from the virus propagation culture at the end of the culturing process.
  • the method according to the invention allows to determine the concentration of virus particles and/or virus antigens in the sample. The concentration can readily be extrapolated to the complete culture volume.
  • the sample comprising the virus particles and/or virus antigens may be derived from an egg culture or from a cell culture, such as a mammalian cell culture.
  • the sample is derived from an MDCK, MDBK, VERO, CV-1, MRC-5, WI-38, or an LLC-MK2 cell culture. According to a even more preferred embodiment, the sample is derived from an MDCK cell culture.
  • the results obtained from the determination method according to the invention can be used as a basis for deciding as to whether a complete virus propagation culture should be further processed and/or purified. Since downstream processing and/or purification of a complete virus propagation culture is extremely cost and labor intensive, the method of the invention provides for a more economic and cost-saving production approach. Cultures which exhibit only a low virus/antigen content at the end of the culturing phase can be discarded and do not have to be subjected to downstream processing and/or purification methods. For example, an acceptable yield of an influenza virus propagation culture can be about 10-20 ⁇ g/ml culture. However, the yield to be expected will differ, dependent on the virus type and the culture method.
  • the determination result can also be the basis for deciding as to whether the virus propagation culture is to be further incubated and/or the culture conditions are to be optimized. It will be understood that in the latter case the aliquot of the virus propagation culture must be obtained under sterile condition.
  • the method for determining the concentration of virus particles and/or virus antigens in a sample further comprises the step of comparing the detection signal to at least one reference value obtained from detecting a sample containing said virus particles and/or virus antigens in a known concentration (reference sample) in order to determine the concentration of virus particles and/or virus antigens in the sample.
  • the specific equipment used for the method of the invention is calibrated prior to applying the sample with the unknown concentration of virus particles and/or virus antigens.
  • the detection signal obtained with the reference sample can be correlated to a specific concentration of virus particles and/or virus antigens.
  • the detection signal is compared to more than one reference value.
  • a calibration curve may be prepared by determining the detection signals of several samples of different known concentrations.
  • the samples may contain between 5 and 50 ⁇ g virus particles and/or virus antigens per ml of the reference sample.
  • reference samples with increasing concentration of virus particles and/or virus antigens are used.
  • samples containing 5 ⁇ g, 10 ⁇ g, 15 ⁇ g, 20 ⁇ g, 25 ⁇ g, 30 ⁇ g, 35 ⁇ g, 40 ⁇ g, 45 ⁇ g, and 50 ⁇ g virus particles and/or virus antigens per ml may be used to generate a calibration curve.
  • the detection signal obtained from a sample of unknown concentration of virus particles and/or virus antigens can then be compared to the calibration curve in order to assess the concentration of virus particles and/or virus antigens.
  • reference values are established for each virus type. It may also be necessary in some cases to establish reference values for different strains of a given virus type (such as in the case of influenza).
  • reference values can be established for the influenza strains A/New Caledonia H1N1, B/Jingsu, A/Wyoming H3N2, A7New York H3N2, B/Malaysia. In several cases it may also be found out that reference values established on the basis of one strain may allow for measuring and assessing samples containing virus particles and/or virus antigens of another strain.
  • Strain specific correlation factors may be established which allow for using such reference values. If the same equipment is used with the same experimental conditions (i.e. the same detector settings, flow rate etc.), it is also possible to use the detector signal (as expressed, for example, by the specific area of a measured peak) as an indicator for the concentration of virus particles and/or virus antigens. In the examples described bellow, the peak area is directly correlated to the concentration of virus particles and/or virus antigens as measured by standard methods (SRD and HA titer, respectively).
  • proteases like trypsin to the infection medium.
  • the proteases extracellularly cleave the precursor protein of haemagglutinin into active haemagglutinin which is involved in the process of cell infection.
  • protease addition does often not lead to reproducible results in every batch.
  • protease addition requires opening the culturing vessel at least once, there is a high risk of contaminating the virus propagation culture. Such contamination, however, would render the virus product obtained from the cell culture unsuitable for subsequent preparation of vaccines.
  • the method allows for assessing the virus propagation culture in terms of quality. For example, if microorganisms other than the virus to be propagated have contaminated the culture, such contamination would be detectable by the method of the invention, either by a low yield of virus or by occurrence of further peaks in the chromatographic analysis.
  • the method according to the invention preferably is for determining the concentration of complete virus particles which are present in a sample.
  • concentration of other virus antigens such as distinct surface proteins. It has been found in course of the invention that the concentration of complete influenza virus particles grown in MDCK cell cultures is low, if the concentration of free (i.e. not virus-associated) influenza virus surface proteins is high. Thus, the concentration of such surface proteins can also be used as an indication as to the yield of viral particles in a sample.
  • a sample derived from a fermenter harvest of a MDCK (Madin-Darby canine kidney) cell culture infected with influenza strain A/Wellington was filtrated using a 0,45 ⁇ m filter before applying to the chromatography column.
  • MDCK Medin-Darby canine kidney
  • a flowrate of 5 ml/min was used.
  • the column was equilibrated for 10 min with washing buffer. Then 50 ml of the sample were injected. The column was washed with 100 ml washing buffer. Elution with 50 ml elution buffer 1 was performed to elute the complete viruses and a further elution step with 50 ml elution buffer 2 was performed to elute viral surface proteins which are not associated with complete virus particles. The latter proteins might be the product of incomplete virus production. Finally the column was rinsed with 50 ml cleaning buffer.
  • Figure 1 shows the absorption spectrum that was obtained for the elution.
  • the peak at 43.53 min represents the virus particle fraction.
  • the peak area reveals that the virus propagation culture is sufficient for further processing of the sample to produce e.g. a vaccine.
  • viral surface proteins which are not associated with complete virus particles can be used as a further indication of the yield of virus particles from the virus propagation culture. The higher the concentration of these unbound viral proteins, the lower the concentration of virus particles is.
  • the area of the peak at 43.53 min should be large, and no peak (or a peak having a small area) should be obtained when elution with 3 M NaCl is performed).
  • Table 1 results of determination of the concentration of virus particles by SRD and analytical CS-chromatography Lot strain SRD value in the primary yield CS Peak area of the virus peak OOCL110503 A/New Caledonia H1N1 10 ⁇ g/ml 282 OOCL110504 A/New Caledonia H1N1 6 ⁇ g/ml 196 OOCC110412 B/Jiangsu 10 ⁇ g/ml 92 OOCC110413 B/Jiangsu 10 ⁇ g/ml 94 OOCB110501 A/Wyoming H3N2 14 ⁇ g/ml 172 OOCY110506 A/New York H3N2 8 ⁇ g/ml 67 OOCY110507 A/New York H3N2 10 ⁇ g/ml 74 OOCY110508 A/New York H3N2 14 ⁇ g/ml 173
  • Table 2 results of determination of the concentration of virus particles by HA titration and analytical CS-chromatography Lot strain HA titer CS Peak area of the virus peak 201005RK-1/24 B/Malaysia 32 26 201005RK-3/24 B/Malaysia 32 19 201005RK-4/24 B/Malaysia 256 41 201005RK-1/48 B/Malaysia 512 93 201005RK-4/48 B/Malaysia 1024 198 100505EV-4/24 A/New York H3N2 128 40 100505EV-3/48 A/New York H3N2 256 44 100505EV-3/72 A/New York H3N2 512 91 100505EV-4/72 A/New York H3N2 1024 119
  • the peak area of the respective virus peak correlates strongly with the concentration of virus particles and/or virus antigens in the sample as determined by common methods.
  • the correlation of the peak area to the concentration values as determined by SRD and HA-titer differs between all tested influenza virus strains.
  • the specific absorption at 280 nm seems to differ between the tested virus strains. It could be assumed that a strain specific correlation factor could compensate this observation.
  • the analytical CS-chromatography can be performed in less than one hour, this method significantly enhances the time-consuming optimizing of the infection process.
  • the analytical CS-chromatography could be used as an in-process-control e.g. to decide the optimal harvesting time of a production scale fermentor.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Urology & Nephrology (AREA)
  • General Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Toxicology (AREA)
  • Biophysics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Description

  • The invention provides a method for determining the concentration of virus particles/virus antigens in a sample. In particular, the invention relates to determining the concentration of influenza virus particles/influenza virus antigens in a sample. The invention relates to the use of a cation exchange matrix for the determination of the concentration of virus particles/virus antigens in a sample.
  • Background of the invention
  • The preparation of vaccines conferring protection against virus-related diseases regularly requires large quantities of virus. Consequently, the respective viruses have to be produced in high yields in a suitable host system. Several methods for the propagation and culturing of viruses have been described in the prior art. Common methods of virus propagating include, for example, culturing the virus in primary cells or in even more complex cell culture systems. For example, influenza vaccines can be derived from virus propagation cultures which make use of embryonated chicken eggs. In these cultures, the virus is isolated from the allantoic fluid of infected eggs and the antigens are used as a vaccine either in the form of complete or disintegrated virus particles. Several viruses, such as influenza, rabies, mumps, measles, rubella and tick-borne encephalitis virus may also be replicated by use of specific mammalian cell lines.
  • These cell cultures allow for economical virus replication and provide a more reliable propagation system than, for example, chicken eggs. Methods for the culturing of viruses in mammalian cells are disclosed, for example in WO 97/37000 .
  • In order to achieve a high virus yield, the host system, culturing conditions and the virus to be propagated must be adapted to one another. Although efforts have been made to optimize distinct propagation systems, the primary virus yield of a virus propagation culture can considerably vary between different batches or production runs. Thus, it is important to provide methods by use of which the virus yield of a certain virus preparation can be determined.
  • Currently, the determination of primary yield of an influenza virus propagation culture is performed, for example, by measuring the haemagglutinin (HA) titer. Here, the aggregation of hen erythrocytes is used to indicate the amount of virus-derived haemagglutinin in the cell-free virus preparations, such as a crude sample aliquot of a given culture. By using increasing dilutions of such samples, an assessment of the HA amount can be made. The amount of haemagglutinin is indicative for the number of viral particles in the sample. However, this method is strain-specific and, moreover, only semiquantitative. In fact, only factor 2-steps in sensitivity can be determined.
  • Alternatively, influenza virus yield can be assessed by employing the well-accepted single radial immunodiffusion (also referred to in literature as SRID or SRD test).
  • This test is described in detail in the European Pharmacopoeia, 5th edition. The test is based on the visible reaction of a virus antigen and its homologeous antibody in a supporting agarose gel. It is performed by incorporating the virus antigen into the gel and allowing antisera to diffuse radially from the points of application through the fixed antigens. Measurable zones resulting from the antigen-antibody complexes can be observed. When the equilibrium between the external and the internal reactant has been established, the circular precipitation area, originating from the site of the external reactant, is directly proportional to the amount of the antigen applied and inversely proportional to the concentration of the antibody in the gel. Standard antigen and standard antibody for use in this test are provided by the British National Institute for Biological Standards and Control (NIBSC), UK. It is to be noted that the SRID test has to be performed over at least two days.
  • US2003/0170710 describes a method of purifying and quantifying adenovirus by anion-exchange chromatography. US4,724,210 teaches a method for the purification of influenza viruses. An important drawback of the above methods as presently used in the field resides in the fact that evaluation of the primary virus yield of a given virus propagation culture according to the above methods can only be reliably performed after having separated the virus from the culture medium. However, purification of virus from large propagation cultures is both cost-intensive and time-consuming. Presently, it is not possible to evaluate the amount of virus in a given virus propagation culture within a period of several hours or less.
  • Thus, methods are needed which rapidly provide information as to the primary yield of a virus preparation, such as an influenza virus propagation culture. Preferably, the determination method should provide a result so rapidly that such result can be used as a basis for deciding whether or not the virus propagation culture is further processed and subjected to elaborate purification methods. The present invention solves this problem and provides additional benefits as well.
  • Detailed Description of the Invention
  • The present invention provides a method for determining the concentration of viral particles and/or antigens in a virus-containing sample within a short period of time as defined in the claims. The virus-containing sample can be a crude sample of a virus propagation culture, such as a culture of MDCK cells, and the method according to the invention is performed with an aliquot of said culture. Advantageously, examination of the aliquot can be performed without the necessity to subject the complete culture volume to downstream purification. Thus, the invention provides a method for determining the concentration of virus particles/virus antigens in a sample comprising the steps of
    • a) contacting a sample containing virus particles and/or virus antigens to a cation-exchange matrix under low salt conditions that allow binding of said virus particles and/or virus antigens to said cation-exchange matrix;
    • b) separating said virus particles and/or virus antigens from other sample components;
    • c) eluting said bound virus particles and/or virus antigens from the matrix; and
    • d. detecting the virus particles and/or virus antigens by measuring UV absorbance of the elution fractions, wherein the detection signal is indicative for the concentration of said virus particles and/or virus antigens in the sample; and
    • e. comparing the detection signal to at least one reference value obtained from detecting a sample containing said virus particles and/or virus antigens in a known concentration in order to determine the concentration of virus particles and/or virus antigens in the sample; wherein the virus is Hepatitis C virus (HCV), measles virus, Hepatitis B virus (HBV), vaccinia virus, lassa virus, Herpes simplex virus 1 (HSV-1), Herpes simplex virus 2 (HSV-2), yellow fever virus, rubella virus, Human immunodeficiency virus (HIV) or influenza virus.
  • In course of the present invention, it has been found that there is a direct correlation between the amount of virus particles and/or virus antigens in a given sample and the amount of virus which can be bound to and eluted from an ion-exchange matrix, preferably a cation exchange matrix. This is surprising, since prior to the present invention such columns have never been contemplated for quantitative analysis of virus particles, such as influenza virus particles.
  • According to the method of the present invention, the virus particles and/or virus antigens in the sample are bound to a cation-exchange matrix in a first step. Such columns can readily be purchased from several manufacturers, such as SP Sepharose™ XL from GE Healthcare Life Sciences, Sartobind S from Sartorius, and the like. According to a preferred aspect of the invention, the column used in the method according to the invention comprises a cellulose matrix.
  • According to a particularly preferred embodiment, the cellulose matrix has a gel exclusion limit of 2000 to 4000 Dalton, more preferably 3000 Dalton. The cellulose matrix preferably comprises activated groups. Preferred activated groups include, for example, sulfate ester or other weak cation exchange groups, such as Carboxymethyl (CM). According to a preferred embodiment, the cellulose matrix comprises sulfate ester as activated groups. Usually, the sulfate esters are present on the cellulose matrix in a comparatively low concentration. For example, the matrix can comprise 500-900, preferably 700 µg sulfur per gram dry matrix material. A cellulose matrix which is particularly suitable for a method according to the invention can be purchased from Millipore GmbH, Eschborn, Germany, under the trademark Matrex® cellufine Sulfate.
  • The sample containing the virus particles and/or virus antigens is contacted with the cation-exchange matrix under low salt conditions that allow binding of said virus particles and/or virus antigens to said cation-exchange matrix. Such conditions are conditions of low ionic strength, i.e. binding of the virus particles and/or virus antigens in step a) is effected under low salt conditions. Preferably, contacting and binding is performed in a suitable buffer solution. Generally, the choice of the buffer is not critical, so that most buffer solutions commonly used for ion-exchange chromatography may be used. Thus, the buffer solution used for contacting the virus particles and/or virus antigens to the matrix can comprise NaCl, Na2HPO4, NaHPO4, Na3PO4, Na3SO4, KCl, K2HPO4, KHPO4, K3PO4, K2SO4, MgCl2, NH4Cl, and the like.
  • The total salt concentration of the sample can be up to 0,1 mol/l, for example, 0,01 mol/l, or 0,05 mol/l. In case the material comprising the virus particles and/or virus antigens (for example an aliquot of a virus propagation culture) has a higher salt concentration, such material can be readily diluted with the above buffer solutions in order to provide a sample of low ionic strength which can be applied in the method of the invention. Furthermore, the material comprising the virus particles and/or virus antigens may be desalted before applying the sample to ion-exchange chromatography. Preferably, contacting of the virus particles and/or virus antigens in step a) is effected in the presence of a buffer solution comprising Na2HPO4. More preferably, the concentration of Na2HPO4 in the sample is between 0,01 and 0,1 mol/l. Most preferably, the concentration of Na2HPO4 in the sample is 0,05 mol/l. Contacting the sample with the matrix can be performed in a pH range of about 6,0 to 8,0, preferably 7,0 to 7,8. Most preferably, contacting is performed at a pH of 7,5. To get quantitative results, one has to ensure that the binding capacity of the respective matrix is by far higher than the amount of virus particles and/or virus antigens in the sample to be examined. If the concentration of virus particles and/or virus antigens is too high, dilution should be considered.
  • After the virus particles and/or virus antigens have been bound to the cation-exchange matrix, several washing steps may be performed in order to separate contaminating components of the sample, for example cellular debris originating from host cells used for virus propagation or medium components, from the virus particles and/or virus antigens.
  • Washing can be performed according to common methods used in the field of chromatography using well known buffers of low ionic strength. For an overview of numerous chromatography methods and techniques, reference is made to "Ion Exchange Chromatography, Principles and Methods", Amersham Pharmacia Edition AA (1998). Preferably, the same buffer which has been used for contacting the virus particles and/or virus antigens to the ion-exchange matrix is used for washing. According to one aspect of the invention, it is preferred to use a sodium phosphate buffer, such as Na2HPO4, having a concentration of 10 to 100 mmol/l for washing. For example, the buffer may have a concentration of 20, 40, 60, 80, or 100 mmol/l. Preferably, the Na2HPO4 buffer has a concentration of 50 mmol/l. As will be appreciated by those skilled in the art, the washing buffer may contain several other components, such as further buffering agents like TRIS (Tris(hydroxymethyl)-aminomethan), MOPS (3-(N-morpholino)-Propansulfonsäure) or HEPES (N-(2-Hydroxyethyl)-piperazin-N'-ethansulfonsäure) or detergents like Triton-X 100, sodium dodecyl sulfate, polysorbate, polyoxyethylene sorbitol esters such as polysorbate 80 (Tween-80) or polysorbate 20 (Tween-20), polyoxypropylene-polyoxyethylene esters such as poloxamer 188, polyoxyethylene alcohols such as BRIJ 35, and the like.
  • Subsequently, the virus particles and/or virus antigens are eluted from the column by use of a buffer containing substances which interfere with the interaction of the virus particles and/or virus antigens with the cation-exchange matrix.
  • Such buffers may comprise the same salts as the buffers used for binding the virus particles and/or virus antigens to the column (see above), however in higher concentrations. Thus, elution of the virus particles and/or virus antigens in step c) is effected under high salt conditions. The concentration of the buffer used for elution depends on the nature of the buffer. For example, the salt concentration of the elution buffer may range from about 0,8 to 5 mol/l. Specifically, the salt concentration of the elution buffer may be 1, 1,2, 1,5, 1,8, 2, 2,2, 2,5, 2,8, 3, 3,5 4, or 4,5 mol/l. Preferably, eluting the virus particles and/or virus antigens in step c) is effected in the presence of a NaCl buffer. The NaCl buffer preferably has a concentration of between 1,0 and 2,0 mol/l, more preferably 1,2 mol/l. Alternatively, an NH4SO4 buffer having a concentration of between 1,0 and 2,0 mol/l can be used for elution. Elution can also be performed in two or more steps using buffers of different concentrations. According to a preferred embodiment of the invention, the elution is performed in two steps, wherein a first elution is performed by use of a sodium chloride buffer of 1,2 mol/l, and subsequently, a second elution is performed by use of a sodium chloride buffer of 3 mol/l. It will be understood by those skilled in the art that elution can also be performed by a linear gradient with increasing salt concentrations.
  • According to the invention, the concentration of both complete virus particles and/or specific parts thereof, such as surface proteins or other protein components (referred to as virus antigens herein), can be determined.
  • As shown in the examples, the method of the invention is particularly suitable to determine the concentration of complete virus particles, such as influenza virus particles, in a sample. Thus, the method provides for determining the yield of a virus propagation culture, such as a MDCK cell culture which has been infected with a virus, such as an influenza virus. Alternatively, if only certain protein components of a virus are produced for subsequent use in vaccines, the present method is also useful for determining their concentration. Moreover, both the concentration of complete virus particles and virus antigens can be determined in parallel, provided that they elute with different retention times.
  • When eluting the virus particles and/or virus antigens, the virus-related substances eluting from the column are detected by measuring the UV absorbance of the eluting substances at a specific wave length. The choice of the wave length depends on the nature of the virus to be detected. For example, virus particles such as influenza virus particles can be detected in a range of 210 to 350 nm, for example 280 nm. According to a preferred aspect of the invention, the eluting substances are detected by absorbance measurement at a wave length of 280 nm. For this purpose, common UV detection means which are well known in the field of chromatography may be used.
  • The virus particles and/or virus antigens to be determined according to the method of the present invention, are HCV, measles virus, HBV, vaccinia virus, lassa virus, HSV-1, HSV-2, yellow fever virus, rubella virus, HIV and influenza virus. Preferably, the virus particles and/or virus antigens originate from an influenza virus. The influenza virus may be selected, for example, from the strains A/New Caledonia H1N1, B/Jingsu, A/Wyoming H3N2, A7New York H3N2, B/Malaysia and other strains. The sample, which comprises the virus particles and/or virus antigens preferably can be an aliquot of a virus propagation culture. As used herein "virus propagation culture" means any culture which serves the purpose of propagating viruses or virus antigens. The term in particular includes any kind of cell or tissue culture, in which the cells or tissues, for example vertebrate cells, are used as a host for the replication of the virus. The aliquot may be taken from the virus propagation culture at the end of the culturing process. The method according to the invention allows to determine the concentration of virus particles and/or virus antigens in the sample. The concentration can readily be extrapolated to the complete culture volume. According to the invention, the sample comprising the virus particles and/or virus antigens may be derived from an egg culture or from a cell culture, such as a mammalian cell culture. According to a preferred embodiment, the sample is derived from an MDCK, MDBK, VERO, CV-1, MRC-5, WI-38, or an LLC-MK2 cell culture. According to a even more preferred embodiment, the sample is derived from an MDCK cell culture.
  • The results obtained from the determination method according to the invention can be used as a basis for deciding as to whether a complete virus propagation culture should be further processed and/or purified. Since downstream processing and/or purification of a complete virus propagation culture is extremely cost and labor intensive, the method of the invention provides for a more economic and cost-saving production approach. Cultures which exhibit only a low virus/antigen content at the end of the culturing phase can be discarded and do not have to be subjected to downstream processing and/or purification methods. For example, an acceptable yield of an influenza virus propagation culture can be about 10-20 µg/ml culture. However, the yield to be expected will differ, dependent on the virus type and the culture method. Alternatively, the determination result can also be the basis for deciding as to whether the virus propagation culture is to be further incubated and/or the culture conditions are to be optimized. It will be understood that in the latter case the aliquot of the virus propagation culture must be obtained under sterile condition.
  • The method for determining the concentration of virus particles and/or virus antigens in a sample further comprises the step of comparing the detection signal to at least one reference value obtained from detecting a sample containing said virus particles and/or virus antigens in a known concentration (reference sample) in order to determine the concentration of virus particles and/or virus antigens in the sample.
  • In other words, the specific equipment used for the method of the invention is calibrated prior to applying the sample with the unknown concentration of virus particles and/or virus antigens. The detection signal obtained with the reference sample can be correlated to a specific concentration of virus particles and/or virus antigens. Preferably, the detection signal is compared to more than one reference value. For example, a calibration curve may be prepared by determining the detection signals of several samples of different known concentrations. The samples may contain between 5 and 50 µg virus particles and/or virus antigens per ml of the reference sample. Typically, reference samples with increasing concentration of virus particles and/or virus antigens are used. For example, samples containing 5 µg, 10 µg, 15 µg, 20 µg, 25 µg, 30 µg, 35 µg, 40 µg, 45 µg, and 50 µg virus particles and/or virus antigens per ml may be used to generate a calibration curve. The detection signal obtained from a sample of unknown concentration of virus particles and/or virus antigens can then be compared to the calibration curve in order to assess the concentration of virus particles and/or virus antigens. Preferably, reference values are established for each virus type. It may also be necessary in some cases to establish reference values for different strains of a given virus type (such as in the case of influenza). For example, reference values can be established for the influenza strains A/New Caledonia H1N1, B/Jingsu, A/Wyoming H3N2, A7New York H3N2, B/Malaysia. In several cases it may also be found out that reference values established on the basis of one strain may allow for measuring and assessing samples containing virus particles and/or virus antigens of another strain.
  • Strain specific correlation factors may be established which allow for using such reference values. If the same equipment is used with the same experimental conditions (i.e. the same detector settings, flow rate etc.), it is also possible to use the detector signal (as expressed, for example, by the specific area of a measured peak) as an indicator for the concentration of virus particles and/or virus antigens. In the examples described bellow, the peak area is directly correlated to the concentration of virus particles and/or virus antigens as measured by standard methods (SRD and HA titer, respectively).
  • When culturing influenza virus in cell cultures, infection of the host cells is dependent on several factors, such as the addition of proteases like trypsin to the infection medium. The proteases extracellularly cleave the precursor protein of haemagglutinin into active haemagglutinin which is involved in the process of cell infection. However, protease addition does often not lead to reproducible results in every batch. Moreover, since protease addition requires opening the culturing vessel at least once, there is a high risk of contaminating the virus propagation culture. Such contamination, however, would render the virus product obtained from the cell culture unsuitable for subsequent preparation of vaccines.
  • According to another aspect described, the method allows for assessing the virus propagation culture in terms of quality. For example, if microorganisms other than the virus to be propagated have contaminated the culture, such contamination would be detectable by the method of the invention, either by a low yield of virus or by occurrence of further peaks in the chromatographic analysis.
  • Such contaminated cultures would be discarded and not subjected to downstream purification. The method according to the invention preferably is for determining the concentration of complete virus particles which are present in a sample. However, it is also possible to determine the concentration of other virus antigens, such as distinct surface proteins. It has been found in course of the invention that the concentration of complete influenza virus particles grown in MDCK cell cultures is low, if the concentration of free (i.e. not virus-associated) influenza virus surface proteins is high. Thus, the concentration of such surface proteins can also be used as an indication as to the yield of viral particles in a sample.
  • Finally, the use of a cation exchange matrix for the determination of the concentration of virus particles and/or virus antigens in a sample is described.
  • Brief description of the figures:
    • Figure 1 shows a chromatogram obtained by running a sample containing influenza virus particles on a Matrex® Cellufine™ sulfate column using the ÄKTAprime chromatographic system. 50 ml fermenter harvest of an influenzs virus, strain A/Wellington, was added to the column. The peak at 43.53 minutes represents the influenza virus particle fraction. As shown in SDS-PAGE analysis, the virus particle fraction exhibited a purity of more than 80% (data not shown).
    EXAMPLES
  • It has to be understood that the following examples merely illustrate the present invention and are not intended to limit the invention as defined in the attached claims.
  • The following equipment was used:
    1. 1. Equipment A
      • ➢ Chromatography system: ÄKTAprime (GE healthcare), 50 ml sample loop, online detection of UV absorbance at 280nm, online detection of conductivity
      • ➢ Column: HR 16/10 (GE healthcare), 20ml Matrex® Cellufine™ sulfate (Millipore)
      • ➢ Washing buffer: 50 mM Na2HPO4, adjusted with 1M NaOH to pH 7,5
      • ➢ Elution buffer 1: 1,2 M NaCl, 50 mM Na2HPO4, adjusted with 1 NaOH to pH 7,5
      • ➢ Elution buffer 2: 3,0 M NaCl, 50 mM Na2HPO4, adjusted with 1 NaOH to pH 7,5
      • ➢ Cleaning buffer: 0,1 NaOH
      • ➢ Storage buffer: Washing buffer + 20% Ethanol
      • ➢ Programming: Flowrate 5ml
        • • 10 min (50ml): column equilibration
        • • 10 min (50ml) : sample injection
        • • 20 min (100ml):column washing with washing buffer
        • • 10 min (50ml): elution with elution buffer 1
        • • 10 min (50ml): elution with elution buffer 2
        • • 10 min (50ml): Cleaning with cleaning buffer
    2. 2. Equipment B
      • ➢ Chromatography system: ÄKTAbasic (GE healthcare), 2 ml sample loop, online detection of UV absorbance at 280nm and 350nm
      • ➢ Column: Tricorn 10/20 (GE healthcare), 2 ml Matrix® Cellufine™ sulfate (Millipore)
      • ➢ Washing buffer: 50 mM Na2HPO4, adjusted with 1M NaOH to pH 7,5
      • ➢ Elutionbuffer : 3,0 M NaCl, 50 mM Na2HPO4, adjusted with 1 M NaOH to pH 7,5
      • ➢ Cleaning buffer: 0,1 M NaOH
      • ➢ Storage buffer: Washing buffer + 20% Ethanol
      • ➢ Programming: Flowrate 5ml
        • • 2 min (10ml): sample injection and column washing with washing buffer
        • • 2 min (10ml): elution with 30% elution buffer, 80% washing buffer
        • • 2 min (10ml): elution with 100% elution buffer
        • • 2 min (10ml): Cleaning with 100% cleaning buffer
        • • 2 min (10ml): column equilibration with washing buffer
    EXAMPLE 1 Chromatographic analysis of a influenza virus sample
  • A sample derived from a fermenter harvest of a MDCK (Madin-Darby canine kidney) cell culture infected with influenza strain A/Wellington was filtrated using a 0,45 µm filter before applying to the chromatography column.
  • A flowrate of 5 ml/min was used. The column was equilibrated for 10 min with washing buffer. Then 50 ml of the sample were injected. The column was washed with 100 ml washing buffer. Elution with 50 ml elution buffer 1 was performed to elute the complete viruses and a further elution step with 50 ml elution buffer 2 was performed to elute viral surface proteins which are not associated with complete virus particles. The latter proteins might be the product of incomplete virus production. Finally the column was rinsed with 50 ml cleaning buffer.
  • Figure 1 shows the absorption spectrum that was obtained for the elution. The peak at 43.53 min represents the virus particle fraction. The peak area reveals that the virus propagation culture is sufficient for further processing of the sample to produce e.g. a vaccine. It was also found out that viral surface proteins which are not associated with complete virus particles can be used as a further indication of the yield of virus particles from the virus propagation culture. The higher the concentration of these unbound viral proteins, the lower the concentration of virus particles is.
  • If the production of complete virus particles is aimed at, the area of the peak at 43.53 min should be large, and no peak (or a peak having a small area) should be obtained when elution with 3 M NaCl is performed).
  • EXAMPLE 2 Correlation of results from chromatographic analysis and SRD analysis of a influenza virus sample
  • Different influenza virus strain were propagated in MDCK cultures according to known methods. At the end of the culturing phase, the concentration of virus particles and/or virus antigens in a 2 ml sample was determined by analytical CS-chromatography (Matrex® Cellufine™ sulfate; Millipore) after filtration using a 0,45 µm filter for removing cellular debris. The established HA titration method was performed in parallel using an equal sample. The confidence limit of the SRD determination is +20%. The results obtained are listed in Table 1. Table 1: results of determination of the concentration of virus particles by SRD and analytical CS-chromatography
    Lot strain SRD value in the primary yield CS Peak area of the virus peak
    OOCL110503 A/New Caledonia H1N1 10 µg/ml 282
    OOCL110504 A/New Caledonia H1N1 6 µg/ml 196
    OOCC110412 B/Jiangsu 10 µg/ml 92
    OOCC110413 B/Jiangsu 10 µg/ml 94
    OOCB110501 A/Wyoming H3N2 14 µg/ml 172
    OOCY110506 A/New York H3N2 8 µg/ml 67
    OOCY110507 A/New York H3N2 10 µg/ml 74
    OOCY110508 A/New York H3N2 14 µg/ml 173
  • EXAMPLE 3 Correlation of the results from chromatographic analysis and HA titration analysis of a influenza virus sample
  • Different influenza virus strain were propagated in MDCK cultures according to known methods. At the end of the culturing phase, the concentration of virus particles and/or virus antigens in a 2 ml sample was determined by analytical CS-chromatography (Matrex® Cellufine™ sulfate; Millipore) after filtration using a 0,45 µm filter for removing cellular debris. The established HA titration method was performed in parallel using an equal sample. The confidence limit of the SRD determination is +20%. The results obtained are listed in Table 2. Table 2: results of determination of the concentration of virus particles by HA titration and analytical CS-chromatography
    Lot strain HA titer CS Peak area of the virus peak
    201005RK-1/24 B/Malaysia 32 26
    201005RK-3/24 B/Malaysia 32 19
    201005RK-4/24 B/Malaysia 256 41
    201005RK-1/48 B/Malaysia 512 93
    201005RK-4/48 B/Malaysia 1024 198
    100505EV-4/24 A/New York H3N2 128 40
    100505EV-3/48 A/New York H3N2 256 44
    100505EV-3/72 A/New York H3N2 512 91
    100505EV-4/72 A/New York H3N2 1024 119
  • It can be inferred from the tables that the peak area of the respective virus peak correlates strongly with the concentration of virus particles and/or virus antigens in the sample as determined by common methods. The correlation of the peak area to the concentration values as determined by SRD and HA-titer differs between all tested influenza virus strains. The specific absorption at 280 nm seems to differ between the tested virus strains. It could be assumed that a strain specific correlation factor could compensate this observation.
  • As the analytical CS-chromatography can be performed in less than one hour, this method significantly enhances the time-consuming optimizing of the infection process. In contrast to the 2-3 day lasting SRD method, the analytical CS-chromatography could be used as an in-process-control e.g. to decide the optimal harvesting time of a production scale fermentor.

Claims (14)

  1. Method for determining the concentration of virus particles and/or virus antigens in a sample comprising the steps of:
    a. applying a sample containing virus particles and/or virus antigens to a cation-exchange matrix under low salt conditions that allow binding of said virus particles and/or virus antigens to said cation-exchange matrix;
    b. separating said virus particles and/or virus antigens from other sample components;
    c. eluting said bound virus particles and/or virus antigens from the matrix;
    d. detecting the virus particles and/or virus antigens by measuring UV absorbance of the elution fractions, wherein the detection signal is indicative for the concentration of said virus particles and/or virus antigens in the sample; and
    e. comparing the detection signal to at least one reference value obtained from detecting a sample containing said virus particles and/or virus antigens in a known concentration in order to determine the concentration of virus particles and/or virus antigens in the sample;
    wherein the virus is Hepatitis C virus (HCV), measles virus, Hepatitis B virus (HBV), vaccinia virus, lassa virus, Herpes simplex virus 1 (HSV-1), Herpes simplex virus 2 (HSV-2), yellow fever virus, rubella virus, Human immunodeficiency virus (HIV) or influenza virus.
  2. Method according to claim 1, wherein the UV absorbance is measured at a wavelength of 210nm-350nm.
  3. Method according to claim 2, wherein the virus particles and/or virus antigens are detected by absorbance measurement at a wavelength of 280nm.
  4. Method according to claim 1, wherein more than one reference value is obtained.
  5. Method according to claim 1, wherein said cation-exchange matrix comprises a cellulose matrix, optionally comprising sulfate ester as activated groups.
  6. Method according to claim 5, wherein said cellulose matrix has a gel exclusion limit of 2000 to 4000 Dalton.
  7. Method according to any of claims 1 to 6, wherein said sample is an aliquot of a virus propagation culture.
  8. Method according to claim 7, wherein said sample is the derived from an egg culture or from a cell culture.
  9. Method according to claim 8, wherein said sample is derived from a MDCK cell culture.
  10. Method according to claims 1-9, wherein contacting of the virus particles and/or virus antigens in step a) is effected in the presence of a buffer comprising Na2HPO4.
  11. Method according to claim 10, wherein the concentration of Na2HPO4 in said buffer is between 0,01 and 0,1 mol/l.
  12. Method according to any of claims 1 to 11, wherein eluting the virus particles and/or virus antigens in step c) is effected under high salt conditions.
  13. Method according to claim 12, wherein eluting the virus particles and/or virus antigens in step c) is effected in the presence of a NaCl buffer.
  14. Method according to claim 13, wherein the concentration of NaCl in said buffer is between 1,0 and 2,0 mol/l.
EP07702744.9A 2006-01-13 2007-01-12 Method for determining the concentration of virus particles/virus antigens Active EP1977245B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP07702744.9A EP1977245B1 (en) 2006-01-13 2007-01-12 Method for determining the concentration of virus particles/virus antigens

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP06000816A EP1808697A1 (en) 2006-01-13 2006-01-13 Use of an ion exchange matrix for determining the concentration of virus particles and/or virus antigens
PCT/EP2007/000272 WO2007080123A1 (en) 2006-01-13 2007-01-12 Method for determining the concentration of virus particles/virus antigens
EP07702744.9A EP1977245B1 (en) 2006-01-13 2007-01-12 Method for determining the concentration of virus particles/virus antigens

Publications (2)

Publication Number Publication Date
EP1977245A1 EP1977245A1 (en) 2008-10-08
EP1977245B1 true EP1977245B1 (en) 2013-04-24

Family

ID=35827699

Family Applications (2)

Application Number Title Priority Date Filing Date
EP06000816A Withdrawn EP1808697A1 (en) 2006-01-13 2006-01-13 Use of an ion exchange matrix for determining the concentration of virus particles and/or virus antigens
EP07702744.9A Active EP1977245B1 (en) 2006-01-13 2007-01-12 Method for determining the concentration of virus particles/virus antigens

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP06000816A Withdrawn EP1808697A1 (en) 2006-01-13 2006-01-13 Use of an ion exchange matrix for determining the concentration of virus particles and/or virus antigens

Country Status (7)

Country Link
US (2) US20090202983A1 (en)
EP (2) EP1808697A1 (en)
JP (1) JP2009523239A (en)
CN (1) CN101395477A (en)
AU (1) AU2007204409B2 (en)
CA (1) CA2636843C (en)
WO (1) WO2007080123A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2933989B1 (en) * 2008-07-16 2013-03-08 Commissariat Energie Atomique PROCESS FOR PURIFYING MICROORGANISMS IN LIQUID SAMPLES
JP5716297B2 (en) 2009-06-25 2015-05-13 Jnc株式会社 Chromatographic packing material, method for producing the same, and method for producing a virus vaccine using the same
EP2966093A1 (en) 2014-07-07 2016-01-13 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Process for the preparation of magnetic sulfated cellulose particles, magnetic sulfated cellulose particles and their use

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4724210A (en) * 1984-08-09 1988-02-09 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Method for purification of influenza virus
WO2002082088A1 (en) * 2001-04-05 2002-10-17 Cavidi Tech Ab Recovery of enzyme activity from enveloped viruses

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2871435B2 (en) * 1993-04-22 1999-03-17 イー・アイ・デュポン・ドウ・ヌムール・アンド・カンパニー Method for producing a hydrophilic polymer coated perfluorocarbon polymer based matrix
SE9702405D0 (en) * 1997-06-24 1997-06-24 Pharmacia Biotech Ab Chromatography materials, a process for their preparation and use of the materials
HUP0102209A2 (en) * 1998-04-22 2001-09-28 Genvec, Inc. Efficient purification of adenovirus
US6689600B1 (en) * 1998-11-16 2004-02-10 Introgen Therapeutics, Inc. Formulation of adenovirus for gene therapy
EP1141249A1 (en) * 1998-12-31 2001-10-10 Aventis Pharma S.A. Method for separating viral particles
AU2003217413A1 (en) * 2002-02-14 2003-09-04 Novavax, Inc. Method for isolation and purification of expressed gene products in vitro
US20040229335A1 (en) * 2003-05-15 2004-11-18 Introgen Therapeutics, Inc. Methods and compositions for the production of adenoviral vectors
JP2005082581A (en) * 2003-09-11 2005-03-31 Masami Moriyama SECRETORY IgA ANTIBODY INDUCER

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4724210A (en) * 1984-08-09 1988-02-09 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Method for purification of influenza virus
WO2002082088A1 (en) * 2001-04-05 2002-10-17 Cavidi Tech Ab Recovery of enzyme activity from enveloped viruses

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
"Methods in Molecular Biology, vol 244, Protein Purification Protocols", 1 January 2004, TOTOWA, NJ, article CHRIS SELKIRK: "Ion-Exchange Chromatography", pages: 125 - 131, XP055039527 *
DEROO T ET AL: "Recombinant neuraminidase vaccine protects against lethal influenza", VACCINE, ELSEVIER LTD, GB, vol. 14, no. 6, 1 April 1996 (1996-04-01), pages 561 - 569, XP004057287, ISSN: 0264-410X, DOI: 10.1016/0264-410X(95)00157-V *
KIYOTAKE TOBITA ET AL: "Spontaneous excretion of virus from MDCK cells persistently infected with influenza virus A/PR/8/34", JOURNAL OF GENERAL VIROLOGY, 1 March 1997 (1997-03-01), pages 563 - 566, XP055021178 *

Also Published As

Publication number Publication date
US20090202983A1 (en) 2009-08-13
EP1808697A1 (en) 2007-07-18
CA2636843A1 (en) 2007-07-19
US20140322700A1 (en) 2014-10-30
CA2636843C (en) 2015-06-02
AU2007204409A1 (en) 2007-07-19
AU2007204409B2 (en) 2013-09-26
JP2009523239A (en) 2009-06-18
WO2007080123A1 (en) 2007-07-19
EP1977245A1 (en) 2008-10-08
CN101395477A (en) 2009-03-25

Similar Documents

Publication Publication Date Title
JP7372392B2 (en) Endotoxin measuring agent
Thompson et al. Analytical technologies for influenza virus-like particle candidate vaccines: challenges and emerging approaches
JP6511685B2 (en) Novel recombinant factor C, method for producing the same, and method for measuring endotoxin
US10415076B2 (en) High throughput quantification and characterization of foot and mouth disease virus and products thereof
Bousse et al. Quantitation of influenza virus using field flow fractionation and multi-angle light scattering for quantifying influenza A particles
EP2022849A1 (en) Method for proliferation of influenza virus
CN104122389B (en) Reagent card for colloidal gold immunochromatograohic assay of lily mottle virus and preparation method of reagent card for colloidal gold immunochromatograohic assay of lily mottle virus
CN108226494B (en) Porcine reproductive and respiratory syndrome virus E L ISA antibody detection kit
CN106872695B (en) A kind of H7 subtype avian influenza virus hemagglutinin antibody indirect ELISA detection kit
Palese et al. Isolation and characterization of influenza virus recombinants with high and low neuraminidase activity: Use of 2-(3′-methoxyphenyl)-N-acetylneuraminic acid to identify cloned populations
US20140322700A1 (en) Method for determining the concentration of virus particles/virus antigens
He et al. Downstream processing of Vero cell-derived human influenza A virus (H1N1) grown in serum-free medium
Zeng et al. Proteomic analysis of SARS associated coronavirus using two-dimensional liquid chromatography mass spectrometry and one-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis followed by mass spectroemtric analysis
CN105349499A (en) Preparation method, product and application of avian influenza whole-virus particle marked vaccine
CN104744590B (en) Anti-H 1 N 1 swine influenza virus hemagglutinin monoclonal antibody and double crush syndrome kit
US20230212230A1 (en) Influenza virus production method using single-use culture process system and rapid confirmation test of influenza virus antigen purification condition
CN101570566B (en) Vero cell cracked protein, preparation method and application thereof
US20120270255A1 (en) Methods for isolating and quantifying antigen from vaccines
JP4108979B2 (en) Biosynthetic carbohydrate deficient transferrin reference standard
US20030124650A1 (en) Method of isolation and purification of trypsin from pronase and use thereof
KR102546626B1 (en) Rapid influenza virus antigen purification condition test
Pepper A chromatographic procedure for the purification of influenza virus
Taha et al. PARTIAL PURIFICATION AND CHARACTERIZATION OF ACID PHOSPHATASE FROM SERA OF OBESE DIABETES MELLITUS PATIENTS
KR20110009210A (en) Lytic proteins of vero cell, preparation method thereof and detection kit for vero hcp comprising the same
CN113848321A (en) Influenza vaccine TPCK-pancreatin residual quantity detection kit

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080724

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

17Q First examination report despatched

Effective date: 20090728

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602007029970

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: G01N0033569000

Ipc: C12Q0001040000

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 7/02 20060101ALI20120914BHEP

Ipc: G01N 33/569 20060101ALI20120914BHEP

Ipc: C12Q 1/04 20060101AFI20120914BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: CH

Ref legal event code: NV

Representative=s name: E. BLUM AND CO. AG PATENT- UND MARKENANWAELTE , CH

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 608687

Country of ref document: AT

Kind code of ref document: T

Effective date: 20130515

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602007029970

Country of ref document: DE

Effective date: 20130620

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130826

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130824

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130725

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130804

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130724

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20140127

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20140114

Year of fee payment: 8

Ref country code: BE

Payment date: 20140114

Year of fee payment: 8

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602007029970

Country of ref document: DE

Effective date: 20140127

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20140109

Year of fee payment: 8

Ref country code: AT

Payment date: 20131224

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140112

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150131

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 608687

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150131

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150131

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150112

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130424

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20070112

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602007029970

Country of ref document: DE

Representative=s name: MUELLER-BORE & PARTNER PATENTANWAELTE PARTG MB, DE

Ref country code: DE

Ref legal event code: R081

Ref document number: 602007029970

Country of ref document: DE

Owner name: SEQIRUS UK LTD., MAIDENHEAD, GB

Free format text: FORMER OWNER: NOVARTIS VACCINES AND DIAGNOSTICS, INC., EMERYVILLE, CALIF., US

REG Reference to a national code

Ref country code: GB

Ref legal event code: 732E

Free format text: REGISTERED BETWEEN 20170831 AND 20170906

REG Reference to a national code

Ref country code: NL

Ref legal event code: PD

Owner name: SEQIRUS UK LIMITED; GB

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), ASSIGNMENT; FORMER OWNER NAME: NOVARTIS VACCINES AND DIAGNOSTICS, INC.

Effective date: 20170906

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 12

REG Reference to a national code

Ref country code: FR

Ref legal event code: TP

Owner name: SEQIRUS UK LIMITED, GB

Effective date: 20171227

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230125

Year of fee payment: 17

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230530

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20240126

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20240129

Year of fee payment: 18

Ref country code: GB

Payment date: 20240129

Year of fee payment: 18